Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Clin Cancer Res ; 28(23): 5190-5201, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36166004

RESUMEN

PURPOSE: Advanced-stage gastrointestinal cancers represent a high unmet need requiring new effective therapies. We investigated the antitumor activity of a novel T cell-engaging antibody (B7-H6/CD3 ITE) targeting B7-H6, a tumor-associated antigen that is expressed in gastrointestinal tumors. EXPERIMENTAL DESIGN: Membrane proteomics and IHC analysis identified B7-H6 as a tumor-associated antigen in gastrointestinal tumor tissues with no to very little expression in normal tissues. The antitumor activity and mode of action of B7-H6/CD3 ITE was evaluated in in vitro coculture assays, in humanized mouse tumor models, and in colorectal cancer precision cut tumor slice cultures. RESULTS: B7-H6 expression was detected in 98% of colorectal cancer, 77% of gastric cancer, and 63% of pancreatic cancer tissue samples. B7-H6/CD3 ITE-mediated redirection of T cells toward B7-H6-positive tumor cells resulted in B7-H6-dependent lysis of tumor cells, activation and proliferation of T cells, and cytokine secretion in in vitro coculture assays, and infiltration of T cells into tumor tissues associated with tumor regression in in vivo colorectal cancer models. In primary patient-derived colorectal cancer precision-cut tumor slice cultures, treatment with B7-H6/CD3 ITE elicited cytokine secretion by endogenous tumor-infiltrating immune cells. Combination with anti-PD-1 further enhanced the activity of the B7-H6/CD3 ITE. CONCLUSION: These data highlight the potential of the B7-H6/CD3 ITE to induce T cell-redirected lysis of tumor cells and recruitment of T cells into noninflamed tumor tissues, leading to antitumor activity in in vitro, in vivo, and human tumor slice cultures, which supports further evaluation in a clinical study.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Gastrointestinales , Ratones , Animales , Humanos , Antígenos B7/metabolismo , Neoplasias Gastrointestinales/tratamiento farmacológico , Linfocitos T , Neoplasias Colorrectales/tratamiento farmacológico , Citocinas , Inmunoglobulina G
2.
Front Immunol ; 13: 1008764, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36159851

RESUMEN

Colorectal cancer (CRC) is one of the most common cancers worldwide and demands more effective treatments. We sought to identify tumor selective CRC antigens and their therapeutic potential for cytotoxic T-cell targeting by transcriptomic and immunohistochemical analysis. LY6G6D was identified as a tumor selectively expressed CRC antigen, mainly in the microsatellite stable (MSS) subtype. A specific anti LY6G6D/CD3 T cell engager (TcE) was generated and demonstrated potent tumor cell killing and T cell activation in vitro. Ex vivo treatment of primary patient-derived CRC tumor slice cultures with the LY6G6D/CD3 TcE led to IFNγ secretion in LY6G6D positive tumor samples. In vivo, LY6G6D/CD3 TcE monotherapy demonstrated tumor regressions in pre-clinical mouse models of engrafted human CRC tumor cells and PBMCs. Lastly, 2D and 3D cocultures of LY6G6D positive and negative cells were used to explore the bystander killing of LY6G6D negative cells after specific activation of T cells by LY6G6D positive cells. LY6G6D/CD3 TcE treatment was shown to lyse target negative cells in the vicinity of target positive cells through a combined effect of IFNγ, TNFα and Fas/FasL. In summary, LY6G6D was identified as a selectively expressed CRC antigen that can be utilized to potently re-direct and activate cytotoxic T-cells to lyse LY6G6D expressing CRC using a TcE. This effect can be spread to target negative neighboring tumor cells, potentially leading to improved therapeutic efficacy.


Asunto(s)
Neoplasias Colorrectales , Factor de Necrosis Tumoral alfa , Animales , Antígenos de Neoplasias , Humanos , Inmunoglobulinas , Activación de Linfocitos , Ratones , Linfocitos T Citotóxicos
4.
Cell Rep ; 25(11): 3074-3085.e5, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30540940

RESUMEN

Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Animales , Antígeno CTLA-4/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Relación Dosis-Respuesta Inmunológica , Resistencia a Antineoplásicos , Hematopoyesis , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/patología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas S100/administración & dosificación , Proteínas S100/inmunología
5.
Am J Respir Crit Care Med ; 197(9): 1164-1176, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29327939

RESUMEN

RATIONALE: C5aR1 (CD88), a receptor for complement anaphylatoxin C5a, is a potent immune mediator. Its impact on malignant growth and dissemination of non-small cell lung cancer cells is poorly understood. OBJECTIVES: To investigate the contribution of the C5a/C5aR1 axis to the malignant phenotype of non-small cell lung cancer cells, particularly in skeletal colonization, a preferential lung metastasis site. METHODS: Association between C5aR1 expression and clinical outcome was assessed in silico and validated by immunohistochemistry. Functional significance was evaluated by lentiviral gene silencing and ligand l-aptamer inhibition in in vivo models of lung cancer bone metastasis. In vitro functional assays for signaling, migration, invasion, metalloprotease activity, and osteoclastogenesis were also performed. MEASUREMENTS AND MAIN RESULTS: High levels of C5aR1 in human lung tumors were significantly associated with shorter recurrence-free survival, overall survival, and bone metastasis. Silencing of C5aR1 in lung cancer cells led to a substantial reduction in skeletal metastatic burden and osteolysis in in vivo models. Furthermore, metalloproteolytic, migratory, and invasive tumor cell activities were modulated in vitro by C5aR1 stimulation or gene silencing. l-Aptamer blockade or C5aR1 silencing significantly reduced the osseous metastatic activity of lung cancer cells in vivo. This effect was associated with decreased osteoclastogenic activity in vitro and was rescued by the exogenous addition of the chemokine CXCL16. CONCLUSIONS: Disruption of C5aR1 signaling in lung cancer cells abrogates their tumor-associated osteoclastogenic activity, impairing osseous colonization. This study unveils the role played by the C5a/C5aR1 axis in lung cancer dissemination and supports its potential use as a novel therapeutic target.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Quimiocina CXCL16/inmunología , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/inmunología , Metástasis de la Neoplasia/inmunología , Receptor de Anafilatoxina C5a/inmunología , Transducción de Señal/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Óseas/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
Adv Exp Med Biol ; 1036: 19-31, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29275462

RESUMEN

Most cancers express tumor antigens that can be recognized by T cells of the host. The fact that cancers become clinically evident nonetheless implies that immune escape must occur. Two major subsets of human melanoma metastases have been identified based on gene expression profiling. One subgroup has a T cell-inflamed phenotype that includes expression of chemokines, T cell markers, and a type I IFN signature. In contrast, the other major subset lacks this phenotype and has been designated as non-T cell-inflamed. The mechanisms of immune escape are likely distinct in these two phenotypes, and therefore the optimal immunotherapeutic interventions necessary to promote clinical responses may be different. The T cell-inflamed tumor microenvironment subset shows the highest expression of negative regulatory factors, including PD-L1, IDO, FoxP3+ Tregs, and evidence for T cell-intrinsic anergy. Therapeutic strategies to overcome these inhibitory mechanisms are being pursued, and anti-PD-1 mAbs have been FDA approved. The presence of multiple inhibitory mechanisms in the same tumor microenvironment argues that combination therapies may be advantageous, several of which are in clinical testing. A new paradigm may be needed to promote de novo inflammation in cases of the non-T cell-infiltrated tumor microenvironment. Natural innate immune sensing of tumors appears to occur via the host STING pathway, type I IFN production, and cross-priming of T cells via CD8α+ DCs. New strategies are being developed to engage this pathway therapeutically, such as through STING agonists. The molecular mechanisms that mediate the presence or absence of the T cell-inflamed tumor microenvironment are being elucidated using parallel genomics platforms. The first oncogene pathway identified that mediates immune exclusion is the Wnt/ß-catenin pathway, suggesting that new pharmacologic strategies to target this pathway should be developed to restore immune access to the tumor microenvironment.


Asunto(s)
Inmunoterapia/métodos , Neoplasias , Linfocitos T , Microambiente Tumoral/inmunología , Animales , Humanos , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Linfocitos T/inmunología , Linfocitos T/patología
7.
Cell Res ; 27(1): 96-108, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27981969

RESUMEN

A pre-existing T cell-inflamed tumor microenvironment has prognostic utility and also can be predictive for response to contemporary cancer immunotherapies. The generation of a spontaneous T cell response against tumor-associated antigens depends on innate immune activation, which drives type I interferon (IFN) production. Recent work has revealed a major role for the STING pathway of cytosolic DNA sensing in this process. This cascade of events contributes to the activation of Batf3-lineage dendritic cells (DCs), which appear to be central to anti-tumor immunity. Non-T cell-inflamed tumors lack chemokines for Batf3 DC recruitment, have few Batf3 DCs, and lack a type I IFN gene signature, suggesting that failed innate immune activation may be the ultimate cause for lack of spontaneous T cell activation and accumulation. With this information in hand, new strategies for triggering innate immune activation and Batf3 DC recruitment are being developed, including novel STING agonists for de novo immune priming. Ultimately, the successful development of effective innate immune activators should expand the fraction of patients that can respond to immunotherapies, such as with checkpoint blockade antibodies.


Asunto(s)
Inmunidad Innata , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Transducción de Señal , Células Dendríticas/inmunología , Humanos , Microbiota
8.
Immunol Rev ; 274(1): 290-306, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27782320

RESUMEN

Chronic inflammation in the tumor microenvironment and evasion of the antitumor effector immune response are two of the emerging hallmarks required for oncogenesis and cancer progression. The innate immune system not only plays a critical role in perpetuating these tumor-promoting hallmarks but also in developing antitumor adaptive immune responses. Thus, understanding the dual role of the innate system in cancer immunology is required for the design of combined immunotherapy strategies able to tackle established tumors. Here, we review recent advances in the understanding of the role of cell populations and soluble components of the innate immune system in cancer, with a focus on complement, the adapter molecule Stimulator of Interferon Genes, natural killer cells, myeloid cells, and B cells.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Proteínas del Sistema Complemento/metabolismo , Inmunidad Innata , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Células Mieloides/inmunología , Neoplasias/inmunología , Inmunidad Adaptativa , Animales , Humanos , Inmunoterapia/tendencias , Neoplasias/terapia , Escape del Tumor , Microambiente Tumoral
9.
J Clin Invest ; 126(7): 2404-11, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27367184

RESUMEN

A major subset of human cancers shows evidence for spontaneous adaptive immunity, which is reflected by the presence of infiltrating CD8+ T cells specific for tumor antigens within the tumor microenvironment. This observation has raised the question of which innate immune sensing pathway might detect the presence of cancer and lead to a natural adaptive antitumor immune response in the absence of exogenous infectious pathogens. Evidence for a critical functional role for type I IFNs led to interrogation of candidate innate immune sensing pathways that might be triggered by tumor presence and induce type I IFN production. Such analyses have revealed a major role for the stimulator of IFN genes pathway (STING pathway), which senses cytosolic tumor-derived DNA within the cytosol of tumor-infiltrating DCs. Activation of this pathway is correlated with IFN-ß production and induction of antitumor T cells. Based on the biology of this natural immune response, pharmacologic agonists of the STING pathway are being developed to augment and optimize STING activation as a cancer therapy. Intratumoral administration of STING agonists results in remarkable therapeutic activity in mouse models, and STING agonists are being carried forward into phase I clinical testing.


Asunto(s)
Interferones/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Inmunidad Adaptativa , Animales , Células Presentadoras de Antígenos/inmunología , Antineoplásicos/farmacología , Linfocitos T CD8-positivos/citología , Citosol/metabolismo , ADN de Neoplasias/análisis , Células Dendríticas/inmunología , Humanos , Inmunidad Innata , Interferón beta/inmunología , Ratones , Microambiente Tumoral/inmunología
10.
Cell Rep ; 15(11): 2357-66, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27264175

RESUMEN

Type I interferon (IFN), essential for spontaneous T cell priming against solid tumors, is generated through recognition of tumor DNA by STING. Interestingly, we observe that type I IFN is not elicited in animals with disseminated acute myeloid leukemia (AML). Further, survival of leukemia-bearing animals is not diminished in the absence of type I IFN signaling, suggesting that STING may not be triggered by AML. However, the STING agonist, DMXAA, induces expression of IFN-ß and other inflammatory cytokines, promotes dendritic cell (DC) maturation, and results in the striking expansion of leukemia-specific T cells. Systemic DMXAA administration significantly extends survival in two AML models. The therapeutic effect of DMXAA is only partially dependent on host type I IFN signaling, suggesting that other cytokines are important. A synthetic cyclic dinucleotide that also activates human STING provided a similar anti-leukemic effect. These data demonstrate that STING is a promising immunotherapeutic target in AML.


Asunto(s)
Inmunidad Innata , Leucemia Mieloide Aguda/inmunología , Proteínas de la Membrana/metabolismo , Transducción de Señal/efectos de los fármacos , Inmunidad Adaptativa/efectos de los fármacos , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ingeniería Genética , Humanos , Inmunidad Innata/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Interferón Tipo I/metabolismo , Leucemia Mieloide Aguda/patología , Ratones Endogámicos C57BL , Análisis de Supervivencia , Xantonas/farmacología
11.
Adv Immunol ; 130: 75-93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26923000

RESUMEN

Despite recent clinical advances in immunotherapy, a fraction of cancer patients fails to respond to these interventions. Evidence from preclinical mouse models as well as clinical samples has provided evidence that the extent of activated T cell infiltration within the tumor microenvironment is associated with clinical response to immunotherapies including checkpoint blockade. Therefore, understanding the molecular mechanisms mediating the lack of T cell infiltration into the tumor microenvironment will be instrumental for the development of new therapeutic strategies to render those patients immunotherapy responsive. Recent data have suggested that major sources of intersubject heterogeneity include differences in somatic mutations in specific oncogene pathways between cancers of individual subjects and also environmental factors including commensal microbial composition. Successful identification of such causal factors should lead to new therapeutic approaches that may facilitate T cell entry into noninflamed tumors and expand the fraction of patients capable of responding to novel immunotherapies.


Asunto(s)
Inmunidad Celular , Inmunoterapia , Neoplasias/terapia , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Animales , Antígenos de Neoplasias/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Inmunidad Celular/genética , Activación de Linfocitos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias Experimentales/inmunología , Resultado del Tratamiento , Vía de Señalización Wnt/inmunología
12.
J Immunol ; 196(7): 3191-8, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26927800

RESUMEN

Recent evidence has indicated that innate immune sensing of cytosolic DNA in dendritic cells via the host STING pathway is a major mechanism leading to spontaneous T cell responses against tumors. However, the impact of the other major pathway triggered by intracellular DNA, the absent in melanoma 2 (AIM2) inflammasome, on the functional output from the stimulator of IFN genes (STING) pathway is poorly understood. We found that dendritic cells and macrophages deficient in AIM2, apoptosis-associated specklike protein, or caspase-1 produced markedly higher IFN-ß in response to DNA. Biochemical analyses showed enhanced generation of cyclic GMP-AMP, STING aggregation, and TANK-binding kinase 1 and IFN regulatory factor 3 phosphorylation in inflammasome-deficient cells. Induction of pyroptosis by the AIM2 inflammasome was a major component of this effect, and inhibition of caspase-1 reduced cell death, augmenting phosphorylation of TANK-binding kinase 1/IFN regulatory factor 3 and production of IFN-ß. Our data suggest that in vitro activation of the AIM2 inflammasome in murine macrophages and dendritic cells leads to reduced activation of the STING pathway, in part through promoting caspase-1-dependent cell death.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/inmunología , ADN/metabolismo , Inflamasomas , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Caspasa 1/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Interferón gamma/biosíntesis , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Nucleotidiltransferasas/metabolismo , Piroptosis/genética , Piroptosis/inmunología
13.
Semin Immunol ; 28(1): 54-63, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26872631

RESUMEN

Immune response to tumors can be successfully oriented for therapeutic purposes, as shown by the clinical efficacy of checkpoint blockade in extending the survival of patients with certain solid and hematologic neoplasms. Nonetheless, numerous patients do not benefit from these new treatments. Tumor-specific CD8(+) T lymphocytes, either endogenously revived by checkpoint interference or adoptively transferred after in vitro expansion and retargeting, can be extremely efficient in controlling metastatic disease but have to overcome a number of restraints imposed by growing tumors. This immune escape relies on a profound modification of the tumor environment, which is rendered less permissive to lymphocyte arrival, persistence, and functional activity. We review here emerging findings on the main negative circuits limiting the efficacy of cancer immunotherapy, as well as novel and conventional approaches that can translate into rational combination therapies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimioradioterapia , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/terapia , Microambiente Tumoral , Animales , Citotoxicidad Inmunológica , Perfilación de la Expresión Génica , Humanos , Terapia de Inmunosupresión , Activación de Linfocitos , Neoplasias/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación
15.
Science ; 350(6264): 1084-9, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26541606

RESUMEN

T cell infiltration of solid tumors is associated with favorable patient outcomes, yet the mechanisms underlying variable immune responses between individuals are not well understood. One possible modulator could be the intestinal microbiota. We compared melanoma growth in mice harboring distinct commensal microbiota and observed differences in spontaneous antitumor immunity, which were eliminated upon cohousing or after fecal transfer. Sequencing of the 16S ribosomal RNA identified Bifidobacterium as associated with the antitumor effects. Oral administration of Bifidobacterium alone improved tumor control to the same degree as programmed cell death protein 1 ligand 1 (PD-L1)-specific antibody therapy (checkpoint blockade), and combination treatment nearly abolished tumor outgrowth. Augmented dendritic cell function leading to enhanced CD8(+) T cell priming and accumulation in the tumor microenvironment mediated the effect. Our data suggest that manipulating the microbiota may modulate cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/inmunología , Bifidobacterium/inmunología , Microbioma Gastrointestinal/inmunología , Melanoma/inmunología , Melanoma/terapia , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/terapia , Animales , Bifidobacterium/genética , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Trasplante de Microbiota Fecal , Regulación de la Expresión Génica , Humanos , Inmunidad/genética , Inmunoterapia/métodos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Simbiosis , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
16.
Clin Cancer Res ; 21(21): 4774-9, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26373573

RESUMEN

Novel immunotherapy approaches are transforming the treatment of cancer, yet many patients remain refractory to these agents. One hypothesis is that immunotherapy fails because of a tumor microenvironment that fails to support recruitment of immune cells, including CD8(+) T cells. Therefore, new approaches designed to initiate a de novo antitumor immune response from within the tumor microenvironment are being pursued. Recent evidence has indicated that spontaneous activation of the Stimulator of Interferon Genes (STING) pathway within tumor-resident dendritic cells leads to type I IFN production and adaptive immune responses against tumors. This pathway is activated in the presence of cytosolic DNA that is detected by the sensor cyclic GMP-AMP synthase (cGAS) and generates cyclic GMP-AMP (cGAMP), which binds and activates STING. As a therapeutic approach, intratumoral injection of STING agonists has demonstrated profound therapeutic effects in multiple mouse tumor models, including melanoma, colon, breast, prostate, and fibrosarcoma. Better characterization of the STING pathway in human tumor recognition, and the development of new pharmacologic approaches to engage this pathway within the tumor microenvironment in patients, are important areas for clinical translation.


Asunto(s)
Inmunoterapia , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/terapia , Transducción de Señal , Animales , Humanos , Inmunidad Innata , Inmunoterapia/métodos , Interferón Tipo I/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/genética , Terapia Molecular Dirigida/métodos , Neoplasias/etiología , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Investigación Biomédica Traslacional , Microambiente Tumoral
17.
Cell Rep ; 11(7): 1018-30, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25959818

RESUMEN

Spontaneous tumor-initiated T cell priming is dependent on IFN-ß production by tumor-resident dendritic cells. On the basis of recent observations indicating that IFN-ß expression was dependent upon activation of the host STING pathway, we hypothesized that direct engagement of STING through intratumoral (IT) administration of specific agonists would result in effective anti-tumor therapy. After proof-of-principle studies using the mouse STING agonist DMXAA showed a potent therapeutic effect, we generated synthetic cyclic dinucleotide (CDN) derivatives that activated all human STING alleles as well as murine STING. IT injection of STING agonists induced profound regression of established tumors in mice and generated substantial systemic immune responses capable of rejecting distant metastases and providing long-lived immunologic memory. Synthetic CDNs have high translational potential as a cancer therapeutic.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias Experimentales/inmunología , Nucleótidos Cíclicos/farmacología , Microambiente Tumoral/inmunología , Animales , Antineoplásicos/síntesis química , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Técnicas de Inactivación de Genes , Humanos , Macrófagos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/tratamiento farmacológico , Nucleótidos Cíclicos/síntesis química , Reacción en Cadena de la Polimerasa , Transfección , Xantonas/farmacología
18.
Front Oncol ; 5: 83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25914882

RESUMEN

Because of its disseminated nature and lack of tumor-draining lymph nodes, acute myeloid leukemia (AML) likely employs unique immune evasion strategies as compared to solid malignancies. Targeting these unique mechanisms may result in improved immunotherapeutic approaches. Emerging data suggest that a specific dendritic cell (DC) subset, CD8α DCs, may be responsible for mediating tolerance in AML and thus targeting the innate immune system may be of benefit in this disease. Promising immune targets include the toll-like receptors, calreticulin/CD47, the stimulator of interferon genes pathway, and signal transducer and activator of transcription 3 (STAT3). However, it is becoming clear that compensatory mechanisms may limit the efficacy of these agents alone and thus rationale combinations of immunotherapies are warranted. This review discusses the potential immune evasion strategies in AML, as well as discussion of the promising innate immune targets, both alone and in combination, for this disease.

19.
Trends Immunol ; 36(4): 250-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25758021

RESUMEN

A major subset of patients with advanced solid tumors show a spontaneous T cell-inflamed tumor microenvironment, which has prognostic import and is associated with clinical response to immunotherapies. As such, understanding the mechanisms governing the generation of spontaneous T cell responses in only a subset of patients is critical for advancing immunotherapeutic approaches further. Here, we discuss the characteristics of T cell-inflamed versus non-inflamed tumors, including a type I interferon (IFN) signature associated with T cell priming against tumor antigens. We review recent findings that have pointed towards the STING (stimulator of interferon genes) pathway of cytosolic DNA sensing as an important innate immune sensing mechanism driving type I IFN production in the tumor context. Knowledge of this pathway is guiding the further development of novel immunotherapeutic strategies.


Asunto(s)
Interferones/inmunología , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T/inmunología , Microambiente Tumoral , Humanos , Interferones/genética
20.
Cytokine Growth Factor Rev ; 26(2): 175-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25630967

RESUMEN

Although type I IFNs were initially described based on their anti-viral properties, it was quickly realized that these cytokines had anti-proliferative and anti-cancer activities. These observations ultimately led to the clinical development and utility of IFN-α2b for the treatment of patients with melanoma, renal cell carcinoma, and chronic myelogenous leukemia, among others. However, the mechanism of action of type I IFNs in vivo was never fully elucidated, and the pleiotropic effects of IFNs on multiple cell types had made it challenging to decipher. Advancement of genetically engineered mouse models has provided new tools for interrogating these mechanisms. Recent evidence has indicated that spontaneous innate immune sensing of cancers that leads to adaptive immune responses is dependent on host type I IFN production and signaling. The major innate immune receptor pathway that leads to type I IFN production in response to a growing tumor appears to be the STING pathway of cytosolic DNA sensing. STING agonists drive type I IFN production and are impressively therapeutic in mouse tumor models. Targeting low doses of type I IFNs to the tumor microenvironment also promotes anti-tumor activity via host adaptive immunity that is T cell-dependent. However, high doses of intratumoral type I IFNs largely function via an anti-angiogenic effect. Understanding these mechanistic details should enable improved clinical manipulation of the type I IFN system in cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón Tipo I/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Inmunidad Adaptativa , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Humanos , Inmunidad Innata , Interferón Tipo I/administración & dosificación , Interferón Tipo I/metabolismo , Ratones , Transducción de Señal , Linfocitos T/inmunología , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA